EP4215609A1 - Polynucléotide pour l'expression physiologique dans des lymphocytes t - Google Patents

Polynucléotide pour l'expression physiologique dans des lymphocytes t Download PDF

Info

Publication number
EP4215609A1
EP4215609A1 EP21868788.7A EP21868788A EP4215609A1 EP 4215609 A1 EP4215609 A1 EP 4215609A1 EP 21868788 A EP21868788 A EP 21868788A EP 4215609 A1 EP4215609 A1 EP 4215609A1
Authority
EP
European Patent Office
Prior art keywords
cells
cell
seq
polynucleotide
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21868788.7A
Other languages
German (de)
English (en)
Inventor
Francisco MARTÍN MOLINA
María TRISTÁN MANZANO
Noelia MALDONADO PÉREZ
Pedro JUSTICIA LIRIO
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fundacion Publica Andaluza Progreso y Salud
Original Assignee
Fundacion Publica Andaluza Progreso y Salud
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from ES202030955A external-priority patent/ES2901575A1/es
Application filed by Fundacion Publica Andaluza Progreso y Salud filed Critical Fundacion Publica Andaluza Progreso y Salud
Publication of EP4215609A1 publication Critical patent/EP4215609A1/fr
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16051Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/15Vector systems having a special element relevant for transcription chimeric enhancer/promoter combination

Definitions

  • the present invention relates to a new technology for generating immunotherapeutic T-cells.
  • the invention provides an improved system for generating immunotherapeutic T-cells comprising a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • Adoptive immunotherapy which involves the transfer of autologous antigen-specific T-cells generated ex vivo, is a promising strategy for treating viral infections and cancer.
  • the T-cells used for adoptive immunotherapy can be generated by means of the expansion of antigen-specific T-cells or the redirection of T-cells by means of genetic engineering.
  • the transfer of viral antigen-specific T-cells is a well-established method which is used for the treatment of transplant-associated viral infections and virus-related rare malignant neoplasms.
  • the isolation and transfer of tumor-specific T-cells have proven successful in the treatment of melanoma.
  • CARs are synthetic receptors consisting of a targeting moiety associated with one or more signaling domains in a single fusion molecule.
  • the binding moiety of a CAR consists of an antigen-binding domain of a single-chain antibody (scFv), comprising lumen 25 and variable fragments of a monoclonal antibody attached together by a flexible linker. Binding moieties based on receptor or ligand domains have also been used successfully.
  • the signaling domains for first-generation CARs are derived from the cytoplasmic region of Fc or CD3zeta receptor gamma chains.
  • First-generation CARs have been proven to successfully redirect T-cell cytotoxicity, however, they failed to provide a prolonged expansion and 30 anti-tumor activity in vivo.
  • the signaling domains of co-stimulatory molecules including CD28, OX-40 (CD134), and 4-1BB (CD137) were added alone (second generation) or in combination (third generation) to improve the survival and increase the proliferation of CAR2-modified T-cells.
  • CARs have allowed T-cells to be successfully redirected against antigens expressed on the surface of tumor cells of different neoplasms, including lymphomas and solid tumors.
  • CD19 has been presented as an attractive immunotherapy target because most B-cell acute lymphoblastic leukemia (B-ALL) 5 uniformly expresses CD19, whereas the expression is absent in non-hematopoietic cells, as well as in myeloid cells, red blood cells, T-cells, and bone marrow stem cells.
  • B-ALL B-cell acute lymphoblastic leukemia
  • Clinical trials targeting CD19 in malignant B-cell tumors are in progress with encouraging anti-tumor responses. Most of them involve the infusion of T-cells genetically modified to express a chimeric antigen receptor (CAR) with specificity derived from the scFv region of a CD19 FMC63-specific mouse monoclonal antibody.
  • CAR chimeric antigen receptor
  • TCR type expression improves the anti-leukemia activity of CAR-T cells by using genome edition systems to express transgenes through the promoter of the TRAC locus.
  • genome edition strategies use highly sophisticated technologies that are hard to implement in clinical practice.
  • the authors of the present invention have developed a system which allows the expression of CAR following the expression pattern of TCR, preventing high expression levels on the surface of T-cells which have been associated with an inefficient long-term therapy or with harmful, life-threatening side effects, such as a cytokine storm.
  • the authors of the invention have selected LV (lentivirus or lentiviral) as the best system to achieve a stable expression and regulatory regions of the B2M locus as a good candidate for expressing CARs which imitate the expression pattern of TCR following CD3/CD28 stimulation, at both protein and mRNA levels.
  • the authors of the invention have created two synthetic promoters including most of the regulatory regions of the B2M locus in a reduced size that will allow insertion thereof in an LV backbone.
  • a first aspect of the invention relates to a polynucleotide comprising or consisting of the sequence SEQ ID NO: 1 or 2, hereinafter polynucleotides of the invention, or a polynucleotide comprising or consisting of a sequence having an identity to SEQ ID NO: 1 or 2 of at least:
  • polynucleotide of the invention of SEQ ID NO 1, also referred to as B2M_EWP1 is a 1161-nucleotide sequence which contains B2 microglobulin (B2M) fragments.
  • polynucleotide of the invention of SEQ ID NO 2, also referred to as B2M_EWP2 is a 1264-nucleotide sequence which contains B2 microglobulin (B2M) fragments.
  • any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) are preferably included in lentiviral vectors for the purpose of enabling the stable expression thereof in cells.
  • a signal sequence (also known as a leader sequence, prepro sequence, or pre-sequence) can be provided in the sequence of any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) or in the sequence of a vector, such as a lentiviral vector, comprising said polynucleotides.
  • the secretory signal sequence will be operatively linked to the transmembrane nucleic acid sequence, i.e., the two sequences bind to the correct reading frame and are positioned to direct the newly synthesized polypeptide into the secretory pathway of the host cell.
  • Secretory signal sequences are usually positioned at 5' with respect to the nucleic acid sequence encoding the polypeptide of interest, although certain secretory signal sequences can be positioned in any other part of the nucleic acid sequence of interest (see, for example, Welch et al., U.S. Patent 5,037,743 ; Holland et al., United States Patent No. 5,143,830 ).
  • secretory signal sequences are usually positioned at 5' with respect to the nucleic acid sequence encoding the polypeptide of interest, although certain secretory signal sequences can be positioned in any other part of the nucleic acid sequence of interest (see, for example, Welch et al., U.S. Patent 5,037,743 ; Holland et al., United States Patent No. 5,143,830 ).
  • Those skilled in the art will recognize that, in view of genetic code degeneration, a considerable sequence variation is possible among these polynucleotide molecules.
  • sequences of any of the polynucleotides of the invention may comprise optimized codons for expression in mammalian cells, preferably for expression in human cells.
  • Codon optimization refers to the exchange, in a sequence of interest, of codons that are generally rare in highly expressed genes of a given species with codons which are generally common in highly expressed genes of such species, such codons encoding the amino acids like the codons being exchanged.
  • another aspect of the invention relates to a gene construct, hereinafter gene construct of the invention, comprising any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2).
  • the gene construct of the invention is a viral vector, and more preferably a lentiviral vector.
  • the gene construct of the invention comprises any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) operatively attached to the sequence of a CAR to drive its expression, wherein the CAR will comprise at least one extracellular ligand-binding domain, a transmembrane domain, and at least one intracellular signaling domain.
  • Another aspect of the invention relates to a cell, hereinafter cell of the invention, comprising any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), or the gene construct of the invention which in turn comprises any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2).
  • the cell of the invention is an immune cell. More preferably, populations of cells of the invention are preferred.
  • the invention relates to a method for the preparation of immune cells for immunotherapy which comprises introducing into said immune cells any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), or the gene construct or vector according to the present invention, and expanding said cells.
  • the invention relates to a method which comprises providing a cell and expressing at least one CAR on the surface of said cell.
  • the method comprises transforming or transducing the cell with at least any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) or with a vector or gene construct comprising any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) operatively attached to the sequence of a CAR, and expressing said polynucleotides in said cell.
  • any of the polynucleotides of the invention is cloned into a vector comprising the CAR of interest.
  • said method further comprises a step of genetically modifying said cell by inactivating at least one gene that expresses a component of the TCR, a target for an immunosuppressive agent, the HLA gene, and/or an immune checkpoint gene such as PD1 or CTLA-4.
  • said gene is selected from the group consisting of TCR-alpha, TCR-beta, CD52, GR, PD1, and CTLA-4.
  • said method further comprises introducing into said T-cells a rare-cutting endonuclease capable of selectively inactivating said genes by means of DNA cleavage.
  • said rare-cutting endonuclease is TALE-nuclease or Cas9 endonuclease.
  • the different methods described above preferably involve the introduction of CAR into a cell using expression vectors comprising or having the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2) cloned therein.
  • said CAR can be introduced as transgenes encoded by a lentiviral vector.
  • the present invention also relates to isolated cells or cell lines which can be obtained by said method for designing cells.
  • said isolated cell comprises at least one CAR and a B2 microglobulin promoter, preferably any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), in particular of SEQ ID NO 1 or 2, operatively attached to CAR to drive its expression.
  • said isolated cell comprises a population of CARs and promoters of the B2 microglobulin locus, in particular of SEQ ID NO 1 or 2, operatively linked to CARs to drive their expression, each comprising different extracellular ligand-binding domains.
  • Immune cells of the present invention are activated and proliferate independently of the antigen binding mechanisms.
  • An isolated immune cell preferably a T-cell obtained according to any of the methods described above, is also included in the scope of the present invention.
  • Said immune cell refers to a cell of hematopoietic origin functionally involved in initiating and/or carrying out an innate and/or adaptive immune response.
  • Said immune cell according to the present invention can derive from a stem cell.
  • the stem cells can be adult stem cells, non-human embryonic stem cells, more particularly non-human stem cells, umbilical cord stem cells, progenitor cells, bone marrow stem cells, induced pluripotent stem cells, totipotent stem cells, or hematopoietic stem cells.
  • Representative human cells are CD34+ cells.
  • Said isolated cell also can be a dendritic cell, a killer dendritic cell, a mast cell, an NK cell, a B-cell, or a T-cell.
  • it is a T-cell selected from the group consisting of inflammatory T lymphocytes, cytotoxic T lymphocytes, regulatory T lymphocytes, or helper T lymphocytes.
  • said cell can derive from the group consisting of CD4+ T lymphocytes and CD8+ T lymphocytes.
  • the cells can be obtained from a series of non-limiting sources, including peripheral blood mononuclear cells, bone marrow, tissue of lymph nodes, umbilical cord blood, tissue of the thymus, tissue of an infection site, ascites, pleural effusion, tissue of the spleen and tumors.
  • any number of T-cell lines available to and known by those skilled in the art can be used.
  • said cell can derive from a healthy donor, a patient diagnosed with cancer, or a patient diagnosed with infection.
  • said cell is part of a mixed population of cells having different phenotype characteristics.
  • the scope of the present invention also encompasses a cell line obtained from a T-cell transformed according to the method described above. Modified cells which are resistant to an immunosuppressive treatment and can be obtained using the preceding method are encompassed in the scope of the present invention.
  • the immune cells can be additionally activated and expanded generally using methods such as those described, for example, in United States Patents 6,352,694 ; 6,534,055 ; 6,905,680 ; 6,692,964 ; 5,858,358 ; 6,887,466 ; 6,905,681 ; 7,144,575 ; 7,067,318 ; 7,172,869 ; 7,232,566 ; 7,175,843 ; 5,883,223 ; 6,905,874 ; 6,797,514 ; 6,867,041 ; and the US patent application with publication no.
  • the T-cells can be expanded in vitro or in vivo.
  • the T-cells of the invention are expanded through contact with an agent that stimulates a CD3/TCR complex and a co-stimulatory molecule on the surface of T-cells to create an activation signal for the T-cells.
  • an agent that stimulates a CD3/TCR complex and a co-stimulatory molecule on the surface of T-cells to create an activation signal for the T-cells.
  • chemicals such as calcium ionophore A23187, phorbol 12-myristate 13-acetate (PMA), or mitogenic lectins such as phytohemagglutinin (PHA)
  • PHA phytohemagglutinin
  • the populations of T-cells can be stimulated in vitro, by contact with an anti-CD3 antibody, or an antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (for example, bryostatin) together with calcium ionophore.
  • a protein kinase C activator for example, bryostatin
  • a ligand which binds to the accessory molecule is used.
  • a population of T-cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody under conditions suitable for stimulating T-cell proliferation.
  • Conditions suitable for culturing T-cells include a suitable medium (for example, minimum essential medium or RPMI1640 medium, or X-vivo 5 (Lonza)) which may contain factors required for proliferation and viability, including serum (for example, human or fetal bovine serum), interleukin-2 (IL-2), insulin, IFN-g, 1L-4, 1L-7, GM-CSF, - 10, -2, 1L-15, TGF, and TNF- or any other cell growth additive.
  • Other cell growth additives include, but are not limited to, surfactant, plasmanate, and reducing agents such as N-acetylcysteine and 2-mercaptoethanol.
  • the media can include RPMI 1640, A1M-V, DMEM, MEM, a-MEM, F-12, X-Vivo 1, and X-Vivo 20, Optimizer, with added amino acids, sodium pyruvate, and vitamins, either without serum or supplemented with a suitable amount of serum (or plasma) or a defined set of hormones, and/or an amount of cytokines sufficient for T-cell growth and expansion.
  • Antibiotics for example, penicillin and streptomycin, are also included in experimental cultures, not in cell cultures that will be infused into a subject.
  • Target cells are kept under conditions required to support growth, for example, at a suitable temperature (for example, 37°C) and atmosphere (for example, air plus 5% CO 2 ). T-cells which have been exposed to several stimulation times may exhibit different characteristics.
  • said cells can be expanded by means of co-culture with tissue or cells. Said cells also can be expanded in vivo, for example, in the blood of the subject after administering said cell to the subject.
  • composition of the invention comprising any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), the gene construct of the invention comprising any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), or the cell of the invention.
  • the composition of the invention further comprises a pharmaceutically acceptable vehicle.
  • the composition of the invention is a pharmaceutical composition.
  • the composition of the invention comprises one or more additional active ingredients.
  • Another aspect of the invention relates to any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), the gene construct of the invention, the cell of the invention, or the composition of the invention, for use thereof in therapy.
  • Another aspect of the invention relates to any of the polynucleotides of the invention (SEQ ID NO 1 or SEQ ID NO 2), the gene construct of the invention, the cell of the invention, or the composition of the invention, for the treatment of cancer.
  • the cancer is selected from the list consisting of neoplasms, B-cell neoplasms, lymphoma, leukemia, and/or myeloma.
  • the isolated cells obtained by the different methods or the cell line derived from said isolated cell as described above can be used as a medication.
  • said medication can be used for the treatment of cancer, particularly for the treatment of B-cell lymphomas and leukemia in a patient in need thereof.
  • said isolated cell according to the invention or cell line derived from said isolated cell can be used in the manufacturing of a medication for the treatment of cancer in a patient in need thereof.
  • the present invention is based on methods for treating patients in need thereof, said method comprising at least one of the following steps:
  • said T-cells of the invention can undergo an in vivo solid T-cell expansion and can persist for a prolonged period of time.
  • Said treatment can be palliative, curative, or prophylactic. It can be part of an autologous immunotherapy or part of an allogenic immunotherapy treatment.
  • Autologous means that the cells, the cell line, or the population of cells used for treating patients originate from said patient. Allogenic is understood to mean that the cells or the population of cells used for treating patients do not originate from said patient, but rather from a donor.
  • Cancers which can be used with the described methods are described in the preceding section.
  • the treatment can be used to treat patients diagnosed with cancer.
  • Cancers which can be treated may comprise non-solid tumors (such as blood tumors, including, among others, pre-B ALL (pediatric indication), adult ALL, mantle cell lymphoma, diffuse large B-cell lymphoma, and the like).
  • Types of cancers to be treated with CARs of the invention include, but are not limited to, certain lymphoid neoplasms or leukemias.
  • Adult tumors/cancers and childhood tumors/cancers are also included.
  • It can be a treatment in combination with one or more cancer therapies selected from the group of antibody therapy, chemotherapy, cytokine therapy, dendritic cell therapy, gene therapy, hormone therapy, laser light therapy, and radiotherapy.
  • said treatment can be administered to patients subjected to an immunosuppressive treatment.
  • the present invention is preferably based on cells or a population of cells, which have been made resistant to at least one immunosuppressive agent due to the inactivation of a gene encoding a receptor for such immunosuppressive agent.
  • the immunosuppressive treatment must help in the selection and expansion of T-cells according to the invention inside the patient.
  • the cells or population of cells according to the present invention can be administered in any convenient manner, even by means of aerosol inhalation, injection, ingestion, transfusion, implantation, or transplant.
  • the compositions described herein can be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, by intramedullary injection, intramuscularly, by intravenous or intralymphatic injection, or intraperitoneally.
  • the cell compositions of the present invention are preferably administered by intravenous injection.
  • the administration of the cells or population of cells can consist of the administration of 10 4 -10 9 cells/kg body weight, preferably 10 5 to 10 6 cells/kg body weight, including all the integer values of numbers of cells within those ranges.
  • the cells or population of cells can be administered in one or more doses.
  • said effective amount of cells is administered as a single dose.
  • said effective amount of cells is administered as more than one dose for a period of time.
  • the time of administration is at the discretion of the attending physician and depends on the clinical condition of the patient.
  • the cells or population of cells can be obtained from any source, such as blood, banks, or a donor, including the patient him/herself. Although individual needs vary, determination of the optimal ranges of the effective amounts of a given type of cell for a particular disease or conditions is within the knowledge of the skilled person.
  • An effective amount means an amount which provides a therapeutic or prophylactic benefit.
  • the administered dose will depend on the age, health, and weight of the intended patient, type of concurrent treatment, if any, the frequency of treatment, and the nature of the desired effect.
  • said effective amount of cells or a composition comprising those cells is administered parenterally.
  • Said administration can be an intravenous administration.
  • Said administration can be performed directly by means of injection into a tumor.
  • the cells are administered to a patient together with (for example, before, simultaneously, or after) any number of forms of treatment including, among others, treatment with agents such as antiviral, cidofovir and interleukin-2, cytarabine (also known as ARA-C), or nataliziimab therapy, treatment for MS patients, or efaliztimab treatment for psoriasis patients, or other treatments for PML patients.
  • agents such as antiviral, cidofovir and interleukin-2, cytarabine (also known as ARA-C), or nataliziimab therapy
  • MS patients or efaliztimab treatment for psoriasis patients, or other treatments for PML patients.
  • the T-cells of the invention can be used in combination with chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate and FK506, antibodies or other immunoablative agents such as CAM PATH, anti-CD3 antibodies, or other antibody, cytoxin, fludaribine, cyclosporin, FK506, rapamycin, mycoplienolic acid, steroid, FR901228 , cytokine, and irradiation therapies.
  • immunosuppressive agents such as cyclosporin, azathioprine, methotrexate, mycophenolate and FK506, antibodies or other immunoablative agents such as CAM PATH, anti-CD3 antibodies, or other antibody, cytoxin, fludaribine, cyclosporin, FK506, rapamycin, mycoplienolic acid, steroid, FR901228 , cytokine, and irradiation therapies.
  • the cell compositions of the present invention are administered to a patient together with (for example, before, simultaneously, or after) a bone marrow transplant, ablative T-cell therapy using chemotherapy agents such as fludarabine, external beam radiotherapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH.
  • chemotherapy agents such as fludarabine, external beam radiotherapy (XRT), cyclophosphamide, or antibodies such as OKT3 or CAMPATH.
  • the cell compositions of the present invention are administered after an ablative B-cell therapy, such as agents which react with CD20, for example, Rituxan.
  • an ablative B-cell therapy such as agents which react with CD20, for example, Rituxan.
  • the subjects can be subjected to a standard high-dose chemotherapy treatment followed by a peripheral blood stem cell transplant.
  • the subjects receive an infusion of expanded immune cells of the present invention after transplant.
  • the expanded cells are administered before or after surgery.
  • nucleic acid or “polynucleotides” refer to nucleotides and/or polynucleotides, such as desoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by polymerase chain reaction (PCR), and fragments generated by ligation, cleavage, endonuclease action, and exonuclease action.
  • Nucleic acid molecules can be made up of monomers which are naturally occurring nucleotides (such as DNA and RNA) or natural nucleotide analogs (e.g., enantiomeric forms of natural nucleotides) or a combination of both.
  • Modified nucleotides can have alterations in sugar and/or pyrimidine or purine base moieties.
  • Sugar modifications include, for example, the replacement of one or more hydroxyl groups with halogens, alkyl groups, amines, and azide groups, or sugars can be functionalized as ethers or esters.
  • the entire sugar fraction can be replaced with sterically and electronically similar structures, such as aza sugars and carbocyclic sugar analogs.
  • Examples of modifications in a base moiety included alkylated purines and pyrimidines, acylated purines or pyrimidines, or other well-known heterocyclic substitutes.
  • Nucleic acid monomers can be attached by means of phosphodiester bonds or analogs of said bonds. Nucleic acids can be single stranded or double stranded.
  • Viral vectors include retrovirus, adenovirus, parvovirus (e.g., adeno-associated viruses), coronavirus, negative-strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive-strand RNA viruses such as picornavirus and alphavirus, and double-stranded DNA viruses including adenovirus, herpesvirus (e.g., herpes simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus), and poxvirus (e.g., vaccinia, fowlpox, and canarypox).
  • orthomyxovirus e.g., influenza virus
  • rhabdovirus e.g., rabies and vesicular stomatitis virus
  • paramyxovirus
  • viruses include Norwalk virus, togavirus, flavivirus, reovirus, papovavirus, hepadnavirus, and hepatitis virus, for example.
  • retroviruses include: avian sarcoma leukosis, mammalian type C virus, type B virus, type D virus, HTLVBLV group, lentivirus, spumavirus ( Coffin, J. M., Retroviridae: The viruses and their replication, In Fundamental Virology, Third Edition, B. N. Fields, et al., Eds., Lippincott-Raven Publishers, Philadelphia, 1996 ).
  • cell lines from the group consisting of CHO-K1 cells; HEK293 cells; Caco2 cells; U2-OS cells; NIH 3T3 cells; NSO cells; SP2 cells; CHO-S cells; DG44 cells; K-562 cells, U-937 cells; MRC5 cells; IMR90 cells; Jurkat cells; HepG2 cells; HeLa cells; HT-1080 cells; HCT-116 cells; Hu-h7 cells; Huvec cells; Muda 4 cells can be selected.
  • All these cell lines can be modified by means of the method of the present invention to provide cell line models to produce, express, quantify, detect, study a gene or a protein of interest; these models can also be used to select biologically active molecules of interest in research and production and in various fields such as chemistry, biofuels, therapeutics, and agronomy, as non-limiting examples.
  • a "co-stimulatory molecule” refers to the cognate binding partner in a T-cell that binds specifically to a co-stimulatory ligand, thereby mediating a co-stimulatory response of the cell such as, but not limited to, proliferation.
  • Co-stimulatory molecules include, but are not limited to, an MHC class I molecule, BTLA, and Toll ligand receptor.
  • a "co-stimulatory signal” refers to a signal which, in combination with primary signal, such as TCR/CD3 ligation, leads to T-cell proliferation and/or upregulation or downregulation of key molecules.
  • the term "subject” or “patient” includes all members of the animal kingdom, including non-human primates and humans.
  • the B2M TetO construct is identical, but with the TetO sequence being placed downstream of TSS, whereas in the B2M Enh5' construct, the position of the enhancer defined by Ensembl was changed from the 3' region to the 5' region.
  • the constructs were reconstituted in 20 ⁇ L of water of ultrapure water under sterility conditions.
  • SEWP is a plasmid which allows the expression of eGFP under the control of SFFV (spleen focus-forming virus) viral promoter.
  • This plasmid (available in the laboratory) was modified to substitute the promoter with chimeric constructs. Combined digestions of all the constructs were performed with BamHl and EcoRI restriction enzymes (New England Biolabs) using buffer 2.1 (New England Biolabs) for 1.5 hours at 37°C. Ultrapure agarose gel electrophoresis was performed and bands were isolated by means of the QIAquick ® Gel Extraction Kit (QIAGEN). At the same time, the SEWP backbone was digested with the same enzymes to obtain the promoter-free fragment.
  • the promoters were ligated with the SEWP fragments using T4 DNA ligase (New England Biolabs) in an insert:vector ratio of 7:1. The reaction was carried out overnight at 16°C. Competent E. coli Stbl3 bacteria (Life Technologies) were transformed with the ligation product, and positive colonies were confirmed by means of a colony PCR, with the following program: 1x (95°C, 10 minutes); 35x (95°C, 30 seconds/62°C, 30 seconds/72°C, 30 seconds); 1x (72°C, 10 minutes), by means of the KAPA Taq PCR kit (Kapa Biosystems) and the following primers (Sigma): Fw-cPPT (5'-ACAGCAGAGATCCAGTTTGG-3') and Rv-eGFP (5'-TCACTCTCGGCATGGACG-3') ( Figure 5). A miniprep using the Wizard ® Plus SV Minipreps DNA Purification System kit (Promega) was performed for ligation-positive colonies, and
  • the plasmids of interest were generated from a third-generation anti-CD19 CAR (Creative Biolabs) having the EF1- ⁇ (Elongation factor 1- ⁇ ) promoter.
  • Four constructs, i.e., CD247, LCK, B2M, and CD4 P1 were cloned into the lentiviral backbone of CAR.
  • digestions with Clal and EcoRI enzymes and buffer 3.1 New England Biolabs
  • Competent bacteria were transformed and positive colonies were again confirmed by means of a colony PCR with primers Fw cPPT-clinical (5'-GTGCAGGGGAAAGAATAGTAG-3') and Rv CD19 (5'-TACAGGACTTTCTTTCTGCC-3').
  • Minipreps for positive colonies were performed with the same kit, and checked by means of Hindlll pattern and sequencing.
  • HEK-293T human embryonic kidney cells, ATCC ® CRL-11268 TM . These are adherent cells which were cultured in T175 flasks with DMEM (Dulbecco's Modified Eagle Medium) medium (Biowest) complemented with 10% fetal bovine serum (FBS) (Gibco) and 1% penicillin/streptomycin (P/S; 0.5% each) (Biowest).
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • P/S penicillin/streptomycin
  • the BxPC-3 line (human pancreatic adenocarcinoma cells, ATCC ® CRL-1687 TM ) is also an adherent cell line which was cultured in T25 flasks with RPMI-1640 (Roswell Park Memorial Institute) medium (Biowest) complemented with 10% FBS and 1% P/S.
  • the Jurkat line acute T-cell leukemia cell line, ATCC ® TIB-152 TM
  • Namalwa line Burkitt lymphoma cell line, ATCC ® CRL-1432 TM
  • are suspended cells which are also cultured with RPMI-1640 medium complemented with 10% FBS and 1% P/S, both cultured in T25 flasks.
  • the four cell lines were kept in incubators at a temperature of 37°C.
  • HEK-293T was kept in an atmosphere with 10% carbon dioxide (CO 2 ), whereas the 3 remaining lines were kept in an atmosphere with 5% CO 2 .
  • the cells were subjected to passage 3 times a week, maintaining a cell density of about 1*10 6 cells/mL.
  • PBMCs peripheral blood mononuclear cells
  • T lymphocytes were isolated from the cell cocktail using the MACSxpress ® Pan T Cell Isolation kit (Miltenyi Biotec), consisting of a mixture of magnetic microsphere-conjugated antibodies targeting most PBMC surface markers with the exception of CD3, so it constitutes a magnetic separation method based on the negative depletion of all cell types with the exception of T-cells (CD3+).
  • MACSxpress ® Pan T Cell Isolation kit MACSxpress ® Pan T Cell Isolation kit (Miltenyi Biotec), consisting of a mixture of magnetic microsphere-conjugated antibodies targeting most PBMC surface markers with the exception of CD3, so it constitutes a magnetic separation method based on the negative depletion of all cell types with the exception of T-cells (CD3+).
  • Isolated T-cells were cultured with TexMACSTM medium (Miltenyi Biotec), a medium specific for T-cells, complemented with 5% human AB serum (Biowest), 1% P/S, and 20 IU/mL of interleukin-2 (IL-2) (Miltenyi Biotec), kept in an incubator at 37°C and 5% CO 2 .
  • T-cells were stimulated through TCR with T cell TransAct TM (Miltenyi Biotec), a polymeric anti-CD3/anti-CD28 nanomatrix. The cells were subjected to passage between 2 and 3 times a week, maintaining a cell density of 1*10 6 cells/mL.
  • Vectors were produced using HEK-293T cells as packaging cells. The cells were seeded into 6-well plates (Life Sciences) with a confluence of more than 90%.
  • a second-generation packaging system which involves the use of 3 lentiviral genome-derived plasmids: 1) Transfer plasmid (B2M-SEWP and B2M-CAR); 2) Packaging plasmid (pCMV8.9) of the HIV virus, and 3) Envelope plasmid (pMD2.G) VSV-G, having the greatest range of infectivity, was used. The ratio of 10:7:3, respectively, was maintained between said plasmids.
  • the chosen transfectant agent was LipoD293TM (SigmaGen Laboratories). Transfection was performed in serum-free DMEM and a medium change was performed with Optimen (Gibco) 5 hours post-transfection to eliminate the long-term toxicity of VSV-G and facilitate subsequent concentration.
  • Viral particles present in the supernatant were collected using sterile 5 mL syringes (Terumo) and filtered using filters with a pore size of 0.45 ⁇ m (Life Sciences). Viral particles were concentrated using 100 kD Amicon Ultra-15 filters (Millipore) by means of centrifugation at 1800 g at 4°C. The vectors were stored at -80°C.
  • Vector titration was performed by calculating efficient particles by means of flow cytometry (FACS Canto II, BD Biosciences).
  • GFP vectors transduction of 5 cell types was performed: Jurkat, Namalwa, HEK-293T, BxPC-3, and primary T-cells.
  • Jurkat and Namalwa cells 100,000 cells were plated per well in 48-well plates and viral vectors were added. To improve transduction efficiency, the cells were subjected to spinoculation. Spinoculation is a centrifugation process at 800 g, 32°C, for 1 hour to favor cell-vector contact. In the case of 293T cells, 100,000 cells were also plated.
  • BxPC-3 cells 50,000 cells were plated per well in a 24-well plate.
  • T-cells 200,000 cells were plated per well in 96-well plates and activated before transduction through TCR using T-cell TransAct TM for 24 hours. Transduction was performed by means of spinoculation. In all the cases, a medium change was performed 5 hours post-transduction and the percentage of transduced T-cells was determined 3 days later by means of flow cytometry.
  • CAR vectors transduction was carried out in Jurkat cells and primary T-cells following the same method.
  • the first step to achieve the present invention was to design promoters that would be used subsequently for regulating the expression of eGFP and CAR.
  • B2M ⁇ -2 microglobulin
  • MHC major histocompatibility complex
  • B2M-derived promoters differ from one another in terms of the presence or absence of the TetO region, as well as the position of the enhancer defined by Ensembl.
  • lentiviral vectors were generated to enable transducing primary T-cells, which will allow generating T-cells that express the eGFP reporter gene under each of the previously designed promoters.
  • Lentiviral vectors were generated following the method explained above, and the titer of the vectors of the invention was calculated in a step prior to T-cell transduction. Once the viral vectors of the invention have been titrated, the same process is followed to transduce primary T-cells previously isolated from a population of T-cells (CD3+) greater than 70%.
  • GFP GFP under the control of B2M-derived promoters
  • both the protein and the messenger RNA were studied. In that sense, GFP and CD3 messenger and protein kinetics were generated.
  • the data was analyzed based on the median fluorescence of both GFP and an anti-CD3 antibody-conjugated fluorochrome at different time points.
  • B2M-derived promoters at the protein level, it can be observed that in B2M Enh5', the expression of GFP duly replicates the physiological expression of CD3. In the case of B2M, the expression of GFP increases in the interval of 0 to 8 hours, unlike in the case of CD3. Interestingly, B2M TetO vectors, in which the only difference with B2M is the insertion of the TetO operon 10 bp from the transcription start site, the expression pattern of GFP is more similar to that of CD3, as said increased expression is observed at 8 hours.
  • the phenotype of the GFP+ T-cells changes over time to a state of high differentiation.
  • the 7-day cell population is still mostly the main memory cell population.
  • the BM2 promoter was cloned into the CAR vector. Once the promoter was cloned into the lentiviral backbone of third-generation CAR and the lentiviral vectors as described in detail in the materials and methods section were generated, T-cells were transduced with CAR vectors.
  • the LTRs allow integration.
  • EGFRt encodes the receptor of truncated epidermal growth factor which allows depleting CAR+ cells, if necessary, (using a monoclonal antibody, cetuximab) and allows indirectly detecting the CAR using a fluorochrome-conjugated anti-EGFRt antibody.
  • T2A is a self-cleaving peptide which allows cutting a long peptide into two short peptides (since CAR and EGFRt are encoded together as a recombinant protein and separated as a result of this mechanism).
  • WPRE is a post-transcriptional regulatory element which enhances both the viral vector titer and the transgene expression.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP21868788.7A 2020-09-21 2021-09-21 Polynucléotide pour l'expression physiologique dans des lymphocytes t Pending EP4215609A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
ES202030954 2020-09-21
ES202030955A ES2901575A1 (es) 2020-09-21 2020-09-21 Polinucleótido para expresión fisiológica en células T
PCT/ES2021/070684 WO2022058640A1 (fr) 2020-09-21 2021-09-21 Polynucléotide pour l'expression physiologique dans des lymphocytes t

Publications (1)

Publication Number Publication Date
EP4215609A1 true EP4215609A1 (fr) 2023-07-26

Family

ID=80775948

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21868788.7A Pending EP4215609A1 (fr) 2020-09-21 2021-09-21 Polynucléotide pour l'expression physiologique dans des lymphocytes t

Country Status (4)

Country Link
US (1) US20240043868A1 (fr)
EP (1) EP4215609A1 (fr)
JP (1) JP2023541705A (fr)
WO (1) WO2022058640A1 (fr)

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
GB8611832D0 (en) 1986-05-15 1986-06-25 Holland I B Polypeptide
US5037743A (en) 1988-08-05 1991-08-06 Zymogenetics, Inc. BAR1 secretion signal
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
MXPA02008265A (es) 2000-02-24 2004-09-10 Xcyte Therapies Inc Concentracion y estimulacion simultanea de calulas.
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
CN110913871B (zh) * 2017-05-17 2024-02-23 西雅图儿童医院(Dba西雅图儿童研究所) 生成哺乳动物t细胞活化诱导型合成启动子(syn+pro)以改善t细胞疗法
CN108018312B (zh) * 2017-12-20 2019-09-10 上海优卡迪生物医药科技有限公司 一种t淋巴细胞白血病的car-t治疗载体及其构建方法和应用
CN110055281B (zh) * 2019-04-25 2020-09-01 山东大学第二医院 一种用于制备car-t的慢病毒载体及其构建方法和应用

Also Published As

Publication number Publication date
JP2023541705A (ja) 2023-10-03
US20240043868A1 (en) 2024-02-08
WO2022058640A1 (fr) 2022-03-24

Similar Documents

Publication Publication Date Title
US11007224B2 (en) CD19 specific chimeric antigen receptor and uses thereof
JP7222954B2 (ja) 癌免疫療法のための栄養膜糖タンパク質(5t4、tpbg)特異的キメラ抗原受容体
EP3194432B1 (fr) Recepteur d'antigene chimere a chaines multiples specifique de ror1
AU2014267436B2 (en) CD19 specific chimeric antigen receptor and uses thereof
ES2876925T3 (es) Receptores de antígeno quiméricos específicos para ROR1 (NTRKR1) para inmunoterapia contra el cáncer
ES2765710T3 (es) Receptores de antígeno quimérico específicos de CD33 para la inmunoterapia del cáncer
EP3174556A1 (fr) Récepteurs d'antigènes chimériques spécifiques de l'egfrviii pour l'immunothérapie du cancer
EP4215609A1 (fr) Polynucléotide pour l'expression physiologique dans des lymphocytes t
US20220009993A1 (en) Polynucleotide for safer and more effective immunotherapies
ES2901575A1 (es) Polinucleótido para expresión fisiológica en células T
AU2015295348B2 (en) ROR1 specific multi-chain chimeric antigen receptor
US20200140560A1 (en) Protease based switch chimeric antigen receptors for safer cell immunotherapy
AU2015295348A1 (en) ROR1 specific multi-chain chimeric antigen receptor
NZ714044B2 (en) Cd19 specific chimeric antigen receptor and uses thereof

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230419

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)